Generic selectors
Exact matches only
Search in title
Search in content
Post Type Selectors
Filter by Categories
Abstracts
Book Review
Case Report
Case Series
CMAS‡ - Pancreas - EUS-FNA Cytopathology (PSC guidelines) S1:1 of 5
CMAS‡ - Pancreas - EUS-FNA Cytopathology (PSC guidelines) S1:3 of 5
CMAS‡ - Pancreas - EUS-FNA Cytopathology (PSC guidelines) S1:4 of 5
CMAS‡ - Pancreas -Sampling Techniques for Cytopathology (PSC guidelines) S1:2 of 5
CMAS‡ - Pancreas- EUS-FNA Cytopathology (PSC guidelines) S1:5 of 5
Commentary
CytoJournal Monograph Related Review Series
CytoJournal Monograph Related Review Series (CMAS), Editorial
CytoJournal Monograph Related Review Series: Editorial
Cytojournal Quiz Case
Editorial
Erratum
Letter to Editor
Letter to the Editor
Letters to Editor
Methodology
Methodology Article
Methodology Articles
Original Article
Pap Smear Collection and Preparation: Key Points
Quiz Case
Research
Research Article
Review
Review Article
Systematic Review and Meta Analysis
View Point
View/Download PDF

Translate this page into:

Research Article
2025
:22;
90
doi:
10.25259/Cytojournal_235_2024

Roxadustat: A catalyst for diabetic wound healing through re-epithelialization and angiogenesis

Department of Burn and Plastic Surgery, General Hospital of Central Theater Command, Wuhan, China
Graduate Joint Training Base of Wuhan University of Science and Technology and Central Theater Command General Hospital, Wuhan, China
School of Medicine, Wuhan University of Science and Technology, Wuhan, China
Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China.
Author image
Qiping Lu
Author image
Kai Xu

*Corresponding authors: Qiping Lu, Department of General Surgery, General Hospital of Central Theater Command, Wuhan, China. lqpresearch@126.com

Kai Xu, Department of Burn and Plastic Surgery, General Hospital of Central Theater Command, Wuhan, China. xukaipro@126.com

Licence
This is an open-access article distributed under the terms of the Creative Commons Attribution-Non Commercial-Share Alike 4.0 License, which allows others to remix, transform, and build upon the work non-commercially, as long as the author is credited and the new creations are licensed under the identical terms.

How to cite this article: Tang D, Lin Q, Xu K, Lu Q. Roxadustat: A catalyst for diabetic wound healing through re-epithelialization and angiogenesis. CytoJournal. 2025;22:90. doi: 10.25259/Cytojournal_235_2024

Abstract

Objective:

Hypoxia-inducible factor 1 (HIF-1) signaling mediates multiple links of wound healing. Tissue hypoxia and dysregulation of HIF-1 signal play a crucial role in non-healing diabetic wounds. Previous studies have found that roxadustat (FG-4592) can promote epidermal stem cell proliferation by upregulating the HIF signaling pathway. This study aimed to investigate the role of roxadustat in the wound healing of diabetic mice.

Material and Methods:

The study was divided into in vivo and in vitro experiments. In the in vivo experiment, mice were categorized into three groups: control group, diabetes group, and diabetes + roxadustat group. Diabetic mice were injected intraperitoneally with roxadustat daily at a dose of 10 mg/kg. Hematoxylin & eosin staining and Masson staining were employed to assess wound healing speed and quality. Immunohistochemical staining was used to detect HIF-1a and proliferating cell nuclear antigens. Western blot was conducted to examine markers associated with Notch1 signaling pathway activation (Notch Intracellular Domain [NICD]), keratinocyte differentiation, and angiogenesis. In the in vitro experiment, HaCaT cells were divided into control (Glu 5.5 mM), high-glucose (Glu 30 mM), and high-glucose + drug (Glu 30 mM + FG-4592) groups, with a treatment concentration of FG-4592 set at 10 µM. Following 48 h of treatment period, protein was extracted for co-immunoprecipitation analysis to determine the interaction between HIF-1a and NICD, and fluorescence staining was conducted to assess their co-localization.

Results:

Roxadustat reversed the slow wound healing caused by diabetes and significantly improved the quality of healing (P < 0.05). It upregulated the inhibited HIF-1 signaling in diabetic mice (P < 0.05) and triggered cell proliferation. It downregulated the hyperactivated Notch1 signaling in diabetic mice (P < 0.05) and induced keratinocyte dedifferentiation, which were both responsible for wound re-epithelialization. Roxadustat also reversed the downregulated expression of vascular endothelial growth factor and CD31 in diabetic mice (P < 0.05) and accelerated the wound angiogenesis process.

Conclusion:

Roxadustat shows potential as a therapeutic drug by promoting re-epithelialization and angiogenesis to bring vigor to the impaired diabetic wound.

Keywords

Angiogenesis inducers
Diabetes complication
Hypoxia-inducible factor 1
Re-epithelialization
Wound healing

INTRODUCTION

The treatment of diabetic wounds remains a major challenge in the field of wound repair. With the advent of the aging society, the incidence of diabetic non-healing wounds is increasing year by year, which brings a heavy burden to the social economy.[1] Compared with the normal wound healing process, the non-healing wound of diabetes showed decreased angiogenesis, decreased recruitment of bone marrow-derived endothelial progenitor cells, and decreased proliferation and migration of fibroblasts and keratinocytes.[2] Obtaining effective treatment strategies targeted for the pathological mechanism of diabetic wounds remains necessary at present.

Wound healing is a dynamic, orderly, and precisely regulated process that requires the coordination of cellular and non-cellular components in space and time.[3] Hypoxia is one of the important microenvironmental factors in the process of wound healing. Hypoxia response is mainly mediated by hypoxia-inducible factor-1 (HIF-1), which has been shown to play a key role in almost all processes of wound healing.[4,5] HIF-1 has two subunits, and HIF-1β protein level is relatively constant. However, under normoxic conditions, HIF-1α is hydroxylated by prolyl hydroxylase and subsequently degraded by the proteasome pathway.[6]

Notably, hyperglycemia can reduce the stability of HIF-1α, which may lead to poor healing and ulcer complications in patients with diabetes.[2,7,8] Diabetic microvascular damage makes the diabetic wound area hypoxic, and high glucose impairs the stability of HIF-1α and the helpful hypoxic response mediated by HIF-1 signaling.[9,10] Hence, the vicious cycle leads to the worsening of severe damage in the wound-healing process. Improving the stability of HIF-1α may be a promising strategy for diabetic non-healing wounds.[8,11]

Roxadustat (FG-4592) is a novel HIF prolyl hydroxylase inhibitor that promotes erythropoiesis and is used to treat renal anemia in clinic.[12,13] The authors’ previous study demonstrated that roxadustat could accelerate wound healing by activating epidermal stem cells through upregulating HIF-1 signaling and promoting the expression of the proliferation marker proliferating cell nuclear antigen (PCNA).[14] A recent study has revealed the effect of roxadustat on angiogenesis through HIF-1α/vascular endothelial growth factor (VEGF) signaling.[15] However, whether roxadustat could reverse the delayed re-epithelialization in diabetic mice, is unclear, and the underlying mechanism associated with reepithelialization needs further study.

Notch1 signaling plays an important role in maintaining skin self-renewal and regulating the proliferation and differentiation of keratinocytes. When Notch1 signaling is activated, it inhibits proliferation and promotes differentiation. Whether roxadustat influences Notch1 signaling, is unknown. Dedifferentiation is an important step in the process of re-epithelization. Among terminally differentiating keratinocytes, some of the surviving keratinocytes reverted from a differentiated to a dedifferentiated state, as evidenced by the re-expression of markers of epidermal stem cells, including integrin β1 and keratin 14 (K14).[16] Dedifferentiation is particularly obvious in split-thickness skin grafting and wound repair of full-thickness skin defects. Whether roxadustat could induce the transformation of terminally differentiated keratinocytes into their precursors in diabetic mice, is currently unknown.

This study aimed to explore the effect and potential mechanism of roxadustat on cutaneous wound healing in diabetic mice. The findings indicated that roxadustat could be a potential therapeutic drug through upregulating HIF signaling and downregulating Notch1 signaling to bring vigor to the impaired diabetic wound. The findings also suggest that roxadustat not only promotes angiogenesis but also accelerates reepithelization in diabetic wounds.

MATERIAL AND METHODS

Ethics statement

This animal experiment followed the requirements of the ethical review system for the experimental animal welfare of Wuhan Myhalic Biotechnology Co., Ltd. Animal Ethics Committee. Approval number HLK-20210906-001. The mice were fed under specific pathogen-free conditions, and they underwent euthanasia after sampling.

Induction of diabetic mice

Male BALB/c mice aged 10–12 weeks (20 ± 2 g) were purchased from Beijing Weitong Lihua Laboratory Animal Technology Co., Ltd (Beijing, China). The mice were certified to be of specific-pathogen-free grade (Animal Quality Certificate Number: No. 110011241101163626).

The mice received a single intraperitoneal injection of streptozotocin (STZ; 160 mg/kg; 18883-66-4, Sigma, USA).[17] After 72 h, their blood glucose level was tested through the tail pricking method, and mice with blood glucose higher than 16.7 mmol/L were recognized as diabetic mice. The diabetes mice were fed for 4 weeks before wound modeling. The healthy mice from the same batch served as a control group.

Full-thickness skin excisional wound model

After anesthesia by intraperitoneal injection of 1% pentobarbital (P5178, Sigma, USA) at 40 mg/kg and hair removal were performed, two full-thickness skin wounds of 4 mm in diameter were made on the back of each mouse as described in the previous study.[14] A self-adhesive silicone ring was placed around the wound to offset wound shrinkage. This silicone inner ring can be a reference for measuring the wound size. Then, all mice were housed individually.

Animal experiment grouping scheme and treatment measures

In accordance with previous studies,[15] roxadustat (S1007, Selleck, USA) was first dissolved in dimethyl sulfoxide at a concentration of 50 mg/ml and further diluted to 1 mg/mL in sterile phosphate-buffered saline (PBS). The mice were randomly divided into three groups, and they received corresponding treatments.

Diabetes + FG-4592 group (n = 6; 12 wounds in total): This group was composed of STZ-induced diabetic mice fed for 4 weeks after successful induction. Roxadustat was injected intraperitoneally (IP) daily at a dose of 10 mg/kg after the wound model was taken.

Diabetes group (n = 6; 12 wounds in total): This group consisted of STZ-induced diabetic mice fed for 4 weeks after successful induction. An equal amount of PBS was injected IP daily after a wound model was established.

Control group (n = 6; 12 wounds in total): This group comprised normal mice from the same batch as the other two groups. An equal amount of PBS was injected IP daily after a wound model was established.

On days 0, 4, 8, and 12 after injury, blood glucose was monitored through the tail vein, and mouse body weight was measured with a balance. Half of the mice (n = 3) in each group were sampled and sacrificed on day 6 after injury; another half (n = 3) was sampled and sacrificed on day 14 after injury. At the end of the experiment, the mice were euthanized by decapitation after pentobarbital anesthesia. The pentobarbital was administered IP at a concentration of 50 mg/mL and a dosage of 150 mg/kg body weight.

Wound area analysis

A camera with a fixed height was used to take pictures to observe the wound-healing process of mice. A silicone ring was used as a scaffold to reduce the effect of rodent skin shrinkage on wound healing. The inner ring of the silicone ring (7 mm in diameter) was innovatively taken as a reference instead of a ruler to avoid stereological differences in observation.

Photographs of the wound were taken every other day until it healed. The inner circle of the silicone splint was taken as a reference. ImageJ (version 1.8) software (National Institutes of Health, USA) was used to track the wound edge, and the actual wound area was calculated. The relative wound area was calculated as the proportion of the actual wound area at a certain time to the original wound area (immediate postoperative wound area).

Histological and immunochemical analysis

Skin samples were collected on days 6 and 14 after surgery. Half of the samples were fixed in 4% paraformaldehyde, and the other half was frozen with liquid nitrogen.

After the fixed samples were gradually dehydrated, they were embedded in paraffin and cut into 4 µm slices perpendicular to the wound surface.

Hematoxylin and eosin (H&E) staining was conducted in accordance with established procedures. Tissue sections were dewaxed, hydrated, and stained with hematoxylin (G1004, Servicebio, China) for nuclear staining. After being rinsed, the sections were stained with eosin (E8090, Solarbio, China) for cytoplasmic and extracellular matrix staining. Subsequently, the sections were dehydrated, cleared, and mounted with a mounting medium.

Masson trichrome staining was performed using a Masson Trichrome Stain Kit (G1340, Solarbio, China) in accordance with the manufacturer’s instructions. Tissue sections were dewaxed, hydrated, and stained with Weigert’s iron hematoxylin for nuclear staining. After being rinsed, the sections were differentiated in acid alcohol and stained with Biebrich scarlet-acid fuchsin for collagen and muscle fiber staining. After being rinsed again, the sections were treated with phosphomolybdic acid to differentiate the fibers. They were then stained with aniline blue for cytoplasm. Finally, the sections were dehydrated, cleared, and mounted.

The stained sections were examined under a light microscope for histological analysis. The thickness of the epidermis and dermis was measured quantitatively by ImageJ (version) 1.8 software (National Institutes of Health, USA).

The expression levels of HIF-1α and PCNA were evaluated by immunochemistry. First, tissue sections were fixed, permeabilized, and blocked. Second, primary antibodies were applied, incubated overnight, washed, and then detected with labeled secondary antibodies. They were washed again, and signals were visualized with a 3,3N-diaminobenzidine tertrahydrochloride horseradish peroxidase color development kit (P0202, Beyotime, China). The dilution ratio of HIF-1α (20960-1-AP, Proteintech, USA) and PCNA (10205-2-AP, Proteintech, USA) was 1:100 and that of horseradish peroxidase (HRP)-labeled Goat Anti-Rabbit immunoglobulin G (H+L; A0208, Beyotime, China) was 1:50. After being mounted, the immunohistochemically stained sections were photographed using an optical microscope. ImageJ (version 1.8) software (National Institutes of Health, USA) was used to measure the mean optical density of HIF-1α and PCNA in new epidermis.

Western blot (WB)

The frozen sample was cut into pieces, grinded in liquid nitrogen, and lysed in radioimmunoprecipitation assay (RIPA) lysis buffer (R0010, Solarbio, Beijing, China) containing protease and phosphatase inhibitor cocktail (P1260, Solarbio, Beijing, China). The protein content was measured using a bicinchoninic acid protein assay kit (PC0020, Solarbio, Beijing, China). An equal amount of protein in each group was separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to polyvinylidene fluoride (PVDF) membranes (YA1701, Solarbio, Beijing, China). The membrane was blocked with 5% skim milk in tris-buffered saline with tween 20 and incubated with the primary antibody overnight. The source and dilution ratio of the antibody was as follows: Integrin β1 (1:1000, 26918-1-AP, Proteintech, Wuhan, China), K14 (1:1000, 22221-1-AP, Proteintech, Wuhan, China), K1 (1:800, 16848-1-AP, Proteintech, Wuhan, China), K10 (1:1000, 18343-1-AP, Proteintech, Wuhan, China), Notch 1 (Notch Intracellular Domain, NICD) (1:2000, 20687-1-AP, Proteintech, Wuhan, China), glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (1:2000, 10494-1-AP, Proteintech, Wuhan, China), β-actin (1:5000, 20536-1-AP, Proteintech, Wuhan, China), VEGF (1:1000, 19003-1-AP, Proteintech, Wuhan, China), and CD31 (1:1000, 28083-1-AP, Proteintech, Wuhan, China). After being washed, the membranes were incubated with HRP-labeled secondary antibodies (1:1000, A0208, Beyotime, China). The membranes were washed again, and signals were visualized with BeyoECL Plus (P0018M, Beyotime, Shanghai, China). Two internal controls, GAPDH and β-actin, were chosen in this experiment. Images were acquired using a gel imaging system, and the obtained images were later analyzed for gray values using ImageJ (version 1.8) software (National Institutes of Health, USA).

Cell experiment grouping scheme and treatment measures

The cell line used in this study was HaCaT (Catalog number: iCell-h066, Supplier: Saibaikang, Shanghai, China). Short tandem repeat profiling was used to verify the identity of the cell line to ensure its authenticity and integrity. Mycoplasma assays were performed to confirm the absence of mycoplasma contamination in the cell lines before their use in the experiments.

HaCaT cells were divided into control (Glu 5.5 mM), high-glucose (Glu30 mM), and high-glucose + drug (Glu30 mM + FG-4592) groups, with a treatment concentration of FG-4592 set at 10 µM. Following 48 h of treatment period, the cells from each group were harvested, and protein was extracted.

Co-immunoprecipitation (Co-IP)

Co-IP was used to clarify the interaction between HIF-1a and NICD. As a positive control group, a certain amount of HaCaT cell protein was collected for WB. HIF-1a antibodies (1:200, sc-13515, Santa Cruz, USA) were pre-processed by mixing with magnetic beads (HY-K0204, MCE, China) for 2 h at 4°C. After the antibodies-and-magnetic beads complex was washed, then a certain amount of HaCaT cell protein was added to them for 2 h at 4°C. After the protein-antibodies-magnetic beads complex was washed, the immunoprecipitated proteins were recovered from the beads by boiling for 5 min in a sample buffer and then analyzed by immunoblotting as previously described.

Immunofluorescence (IF)

Cells were seeded onto coverslips and grown to an appropriate density. After being washed with PBS, the cells were fixed with 4% paraformaldehyde at room temperature for 15 min. They were then permeabilized with 0.1% Triton X-100 for 5 min, followed by washing with PBS. Afterward, the cells were blocked with 5% BSA for 30 min. The primary antibodies for HIF-1α (1:100, 66730-1-Ig, Proteintech, China) and Notch1 ICD (1:100, PAB35376, Bioswamp, China) were incubated overnight at 4°C. After the cells were washed with PBS, Alexa Fluor 594-conjugated Goat Anti-Mouse (1:200, SAB51359, Bioswamp, Wuhan, China) and HyperFluor 488-conjugated Goat anti-Rabbit (1:200, SAB43742, Bioswamp, Wuhan, China) were added, and the cells were incubated in the dark at room temperature for 1 h. After being washed with PBS, the cells were stained with DAPI (C0065, Solarbio, Beijing, China) and mounted. Fluorescent signals were observed using a fluorescence microscope (Leica DFC7000T, Germany).

Statistical analysis

Statistical comparisons and drawing were conducted in GraphPad Prism (version 6.0) software (GraphPad Software, San Diego, CA, USA). All data were first subjected to tests for normality and homogeneity of variances. Then, data that conformed to a normal distribution by the Shapiro-Wilk test and had homogeneous variances were presented as mean ± standard deviation. One-way analysis of variance (ANOVA) was used for the comparison between groups, and Fisher’s Least Significant Difference test was used for post-test and multiple comparisons of one-way ANOVA. All statistical tests were two-sided, with a P < 0.05 considered statistically significant. Fig Draw software (version 2.0, www.figdraw.com) was used for data visualization.

RESULTS

Roxadustat (FG-4592) accelerates the delayed wound healing of diabetic mice

The speed of wound closure is the most important parameter when evaluating the effect of roxadustat (FG-4592) on diabetic wound healing. As shown in Figures 1a and b, the relative wound area among all groups (P < 0.05) on days 4, 8, and 12 had significant differences.

Roxadustat (FG-4592) accelerates the delayed wound healing of diabetic mice. (a) Photos of wounds in each group. (b) Relative wound area of each group. (c) Blood glucose of each group. (d) Body weight of each group. (e) Weight change compared with the weight at the beginning of diabetes model induction in each group. n = 6 in each group. ✶P < 0.05.
Figure 1:
Roxadustat (FG-4592) accelerates the delayed wound healing of diabetic mice. (a) Photos of wounds in each group. (b) Relative wound area of each group. (c) Blood glucose of each group. (d) Body weight of each group. (e) Weight change compared with the weight at the beginning of diabetes model induction in each group. n = 6 in each group. P < 0.05.

First, the relative wound area of the diabetes group was significantly larger than that of the control group on days 4 (P < 0.05), 8 (P < 0.05), and 12 (P < 0.05) after injury. This finding indicated that the diabetic wound modeling was successful because this model clearly demonstrated the delayed healing of diabetic wounds. The relative wound area of the diabetes + FG-4592 group was significantly smaller than that of the diabetes group on days 4 (P < 0.05), 8 (P < 0.05), and 12 (P < 0.05) after injury, indicating that FG-4592 accelerated the delayed wound healing of diabetic mice.

Blood glucose was continuously monitored during the healing process. As shown in Figure 1c, the blood glucose level of the control group fluctuated between 5 and 11 mmol/L, whereas those of diabetes and diabetes + FG-4592 groups were stably higher than 16.7 mmol/L. No significant difference was found between the two groups, indicating that the diabetes model was stable and reliable and that FG-4592 had no effect on blood glucose.

The weight of the mice was continuously monitored before and after diabetes induction and during wound healing. As shown in Figure 1d, the body weight of the control group was higher than that of the diabetes and diabetes + FG-4592 groups (P < 0.05). The weight change in each group was compared with the weight at the beginning of diabetes induction, as shown in Figure 1e. The weight of the control group increased steadily. Conversely, the weight of the diabetes and diabetes + FG-4592 groups clearly decreased compared with that at the beginning of the diabetes induction. In the daily management of mice, the diabetic mice demonstrated obvious symptoms of polydipsia, polyphagia, and polyuria.

Roxadustat (FG-4592) improves the wound-healing quality of diabetic mice

H&E staining and Masson staining were applied to evaluate the healing quality. As shown in Figures 2a and b, in comparison with the control group, the newly healed skin of the diabetes group in the epidermis and dermis was thin, with few blood vessels, lessened collagen deposition, and reduced wound contraction. Meanwhile, the diabetes + FG-4592 group had an irregularly thickened epidermal layer, more collagen deposition, and more considerable wound contraction than the diabetes group. These findings are consistent with the phenomenon shown in Figure 1a. Quantitative analysis showed that the newly healed skin of normal mice (451.4 ± 47.80µm) was the thickest, followed by the diabetes + FG-4592 group (286.2 ± 79.99µm), whereas that of the diabetes group (151.9 ± 26.09 µm) was the thinnest [Figure 2c]. Significant differences were found in the thickness of newly healed skin among all groups. The skin of the diabetes group was significantly thinner than that of the control group (P < 0.05) and the diabetes + FG-4592 group (P < 0.05).

Roxadustat (FG-4592) improves the wound healing quality of diabetic mice. H&E staining (a) and Masson staining (b) of wound on day 14 after surgery. Bar = 500 μm. Images in the first and second rows are in the same group and both on day 14 after surgery, yellow arrows indicate epidermis, and white arrows indicate dermis. (c) Skin thickness of healed skin in each group at day 14 after surgery. n = 6 (3 from H&E staining sections and 3 from Masson staining sections). ✶P < 0.05. H&E: Hematoxylin and eosin.
Figure 2:
Roxadustat (FG-4592) improves the wound healing quality of diabetic mice. H&E staining (a) and Masson staining (b) of wound on day 14 after surgery. Bar = 500 μm. Images in the first and second rows are in the same group and both on day 14 after surgery, yellow arrows indicate epidermis, and white arrows indicate dermis. (c) Skin thickness of healed skin in each group at day 14 after surgery. n = 6 (3 from H&E staining sections and 3 from Masson staining sections). P < 0.05. H&E: Hematoxylin and eosin.

Roxadustat (FG-4592) upregulates HIF-1 signaling and reverses the depressed proliferation of keratinocytes in diabetic mice

Promoting cell proliferation is the key to accelerate wound healing. Immunohistochemical staining was performed to confirm the effect of roxadustat (FG-4592) on HIF-1 signaling and cell proliferation. As shown in Figure 3a, in the diabetes + FG-4592 group, the HIF-1α highly-expressed region was mainly concentrated in the irregular thickening area of the epidermis. In the control and diabetes groups, the brown areas were relatively few and scattered in the epidermis. HIF-1α was expressed in dermal fibroblasts, but the difference was not as pronounced as in the epidermis.

Roxadustat (FG-4592) upregulates HIF-1 signaling and reverses the depressed proliferation of keratinocytes in diabetic mice. Immunohistochemical image of (a) HIF-1α and (b) PCNA on day 14 after surgery; bar = (a) 200 μm and (b) 500 μm for first row. The image in the second row is an enlarged version of the area in the black box in the first row; bar = 50 μm in the enlarged picture. Mean optical density of HIF-1α (c) and PCNA (d) in the epidermis layer. n = 3 wound samples in each group. ✶P < 0.05. HIF-1: Hypoxia-inducible factor 1, PCNA: Proliferating cell nuclear antigen.
Figure 3:
Roxadustat (FG-4592) upregulates HIF-1 signaling and reverses the depressed proliferation of keratinocytes in diabetic mice. Immunohistochemical image of (a) HIF-1α and (b) PCNA on day 14 after surgery; bar = (a) 200 μm and (b) 500 μm for first row. The image in the second row is an enlarged version of the area in the black box in the first row; bar = 50 μm in the enlarged picture. Mean optical density of HIF-1α (c) and PCNA (d) in the epidermis layer. n = 3 wound samples in each group. P < 0.05. HIF-1: Hypoxia-inducible factor 1, PCNA: Proliferating cell nuclear antigen.

As shown in Figure 3b, in the diabetes + FG-4592 group, the PCNA+ cells were densely distributed in the newly healed epidermis, almost covering all layers of the epidermis. In the diabetes group, the PCNA+ cells were the fewest and scattered in the already thin epidermis. In the control group, the epidermis thickness was uniform, and the PCNA+ cells were mainly distributed in the basal layer. The immunohistochemical images appeared to exhibit overdevelopment, often resulting in excessively dark staining. While PCNA is primarily expressed in the cell nucleus, a notably darkened staining was observed in the cytoplasm of the diabetes + FG-4592 group, and this finding may be attributed to edge effects due to overdevelopment.

As shown in Figures 3c and d, the expression of levels HIF-1α and PCNA significantly differed among groups. The HIF-1α expression in the diabetes group was significantly lower than that in the control group (P < 0.05) and diabetes + FG-4592 group (P < 0.05). Similarly, the PCNA expression in the diabetes group was significantly lower than that in the control group (P < 0.05) and diabetes + FG-4592 group (P < 0.05).

Roxadustat (FG-4592) promotes dedifferentiation of keratinocytes and angiogenesis in diabetic mice

The expression levels of integrin β1, K14, K10, K1, Notch1 NICD, CD31, and VEGF were evaluated by WB to investigate the effect of roxadustat (FG-4592) on keratinocyte differentiation and angiogenesis in diabetic wounds.

As shown in Figure 4a-4f, the expression levels of K14 and integrin β1, markers of basal cells, in the diabetes group were significantly reduced compared with those in the control group (P < 0.05). The expression of K10, the marker of differentiated cells, in the diabetes group significantly increased (P < 0.05), whereas that of K1 decreased (P < 0.05). The treatment of FG-4592 in diabetic mice induced keratinocytes to remain in an undifferentiated state (K14+ and integrin β1+) in diabetic wounds. The activation of Notch1 signaling was detected to understand the underlying mechanisms. The diabetic group demonstrated hyperactivation of Notch1 signaling compared with the control group (P < 0.05). Meanwhile, the FG-4592-treated group exhibited downregulation of Notch1 signaling compared with the diabetes group (P < 0.05). The trend was almost the same on days 6 and 14 post-injury. The change trends of K1 and K10 were not the same, which needs further investigation.

Roxadustat (FG-4592) promotes dedifferentiation of keratinocytes and angiogenesis in diabetic mice. (a) Expression levels of integrin β1, K14, K10, K1, and Notch1 NICD evaluated by Western blot in the middle and at the end of wound healing. (b-f) Corresponding quantitative analysis. (g) CD31 and VEGF expression levels evaluated by Western blot. (h and i) Corresponding quantitative analysis. n = 3 in each group. ✶P < 0.05. NICD: Notch Intracellular Domain, VEGF: Vascular endothelial growth factor, K14: Keratin 14, K10: Keratin 10, K1: Keratin 1.
Figure 4:
Roxadustat (FG-4592) promotes dedifferentiation of keratinocytes and angiogenesis in diabetic mice. (a) Expression levels of integrin β1, K14, K10, K1, and Notch1 NICD evaluated by Western blot in the middle and at the end of wound healing. (b-f) Corresponding quantitative analysis. (g) CD31 and VEGF expression levels evaluated by Western blot. (h and i) Corresponding quantitative analysis. n = 3 in each group. P < 0.05. NICD: Notch Intracellular Domain, VEGF: Vascular endothelial growth factor, K14: Keratin 14, K10: Keratin 10, K1: Keratin 1.

As shown in Figure 4g-4i, the expression levels of VEGF and CD31 in the diabetes group notably decreased in comparison with those of the control group (P < 0.05). Compared with the diabetes group, the FG-4592-treated group showed a significant increase in the VEGF and CD31 expression levels (P < 0.05).

Roxadustat (FG-4592) promotes dedifferentiation in keratinocytes through interaction between HIF-1a and NICD

HaCaT cells were treated with 30 mM glucose for 48 h to simulate high glucose in vitro to further validate the dedifferentiation effect of FG-4592 on keratinocytes. The reversal effect of 10 µM FG-4592 on high-glucose-induced Notch signaling overactivation and keratinocyte over-differentiation was detected. As shown in Figures 5a and b, IF and WB showed that compared with the control group, the high-glucose group had decreased expression levels of HIF-1a, integrin β1, and K14 and an increased expression of Notch1 NICD. Compared with the high-glucose group, the high-glucose + drug group showed an increase in the expression levels of HIF-1a, integrin β1, and K14 and a decrease in the expression of Notch1 NICD. The differences were statistically significant according to quantitative analysis through WB [Figure 5c-f].

Roxadustat (FG-4592) promotes dedifferentiation through interaction between HIF-1α and NICD. (a) Immunofluorescence showing the expression and co-localization of HIF-1α and NICD in HaCaT cells under different conditions; bar = 100 μm. (b) Western blot showing the expression levels of HIF-1α, NICD, K14, and integrin-β1 in HaCaT cells under different conditions. (c-f) Quantitative analysis of WB. n = 3 in each group; ✶P < 0.05. (g) Co-IP confirming the interaction between HIF-1α and NICD. HIF-1: Hypoxia-inducible factor 1, NICD: Notch intracellular domain, WB: Western blot, Co-IP: Co-immunoprecipitation, K14: Keratin 14.
Figure 5:
Roxadustat (FG-4592) promotes dedifferentiation through interaction between HIF-1α and NICD. (a) Immunofluorescence showing the expression and co-localization of HIF-1α and NICD in HaCaT cells under different conditions; bar = 100 μm. (b) Western blot showing the expression levels of HIF-1α, NICD, K14, and integrin-β1 in HaCaT cells under different conditions. (c-f) Quantitative analysis of WB. n = 3 in each group; P < 0.05. (g) Co-IP confirming the interaction between HIF-1α and NICD. HIF-1: Hypoxia-inducible factor 1, NICD: Notch intracellular domain, WB: Western blot, Co-IP: Co-immunoprecipitation, K14: Keratin 14.

Co-localization of HIF-1a and Notch1 NICD [Figure 5a] was observed in IF staining and co-immunoprecipitation further confirmed the interaction between HIF-1a and Notch1 NICD [Figure 5g].

DISCUSSION

In this study, an STZ-induced diabetic mice model was constructed, and the wound-healing process was compared. The results showed that roxadustat could accelerate diabetic wound healing and improve healing quality. Roxadustat also upregulated the depressed HIF-1 signaling in diabetic wounds and downregulated the hyperactivated Notch1 signaling. While the above findings were further verified in the cell experiments, the results found that roxadustat reversed the excessive differentiation of keratinocytes under high-glucose conditions, and this phenomenon may be related to the interaction between HIF-1a and Notch1 NICD.

Previous studies have mainly focused on the effect of roxadustat on angiogenesis.[15,18,19] The present study focused on the reversal effect of roxadustat on the “high differentiation and low proliferation” state of keratinocytes in the context of diabetes [Figure 6]. Therefore, the beneficial effect of roxadustat on diabetic wound healing is reflected not only in accelerating vascularization but also in accelerating re-epithelialization.

Schematic model of roxadustat reversing the “high differentiated and low proliferation” state of keratinocytes in diabetic wounds. In normal skin, keratinocytes maintain a relatively stable rhythm of proliferation and differentiation. Injury can activate the process of wound repair, upregulate HIF-1 signal, downregulate Notch1 signal, and make keratinocytes in a state of “high proliferation and low differentiation.” In the context of diabetes, the skin is usually thin and wound healing is delayed, HIF-1 signaling is inhibited, and Notch1 signaling is continuously activated, making keratinocytes in a state of “low proliferation and high differentiation.” FG-4592 upregulates the inhibited HIF-1 signaling and downregulates the hyperactivated Notch1 signaling, which both benefit diabetic wound re-epithelialization. By Fig draw (version 2.0, www.figdraw.com). HIF-1: Hypoxia-inducible factor 1.
Figure 6:
Schematic model of roxadustat reversing the “high differentiated and low proliferation” state of keratinocytes in diabetic wounds. In normal skin, keratinocytes maintain a relatively stable rhythm of proliferation and differentiation. Injury can activate the process of wound repair, upregulate HIF-1 signal, downregulate Notch1 signal, and make keratinocytes in a state of “high proliferation and low differentiation.” In the context of diabetes, the skin is usually thin and wound healing is delayed, HIF-1 signaling is inhibited, and Notch1 signaling is continuously activated, making keratinocytes in a state of “low proliferation and high differentiation.” FG-4592 upregulates the inhibited HIF-1 signaling and downregulates the hyperactivated Notch1 signaling, which both benefit diabetic wound re-epithelialization. By Fig draw (version 2.0, www.figdraw.com). HIF-1: Hypoxia-inducible factor 1.

Wound healing involves multiple processes, including inflammation, angiogenesis, collagen deposition, and reepithelialization, all of which are finely regulated by the hypoxic response.[4] In diabetes, multiple tissues are in a hypoxic state, and this hypoxic state is difficult to correct due to the downregulation of HIF signaling in hyperglycemia.[2] Previous studies have found the pathogenic role of hypoxia and dysregulated HIF signaling in the development of diabetes and diabetic complications.[20] In recent years, an increasing number of researchers have focused on the strategies to stabilize HIF-1α to improve diabetic wound healing.[21,22]

The study on the effect of roxadustat on diabetic wounds has substantial clinical translational potential. Zhu et al.[15] reported that roxadustat promotes angiogenesis. However, the function of roxadustat on re-epithelialization in diabetic wound healing has not been fully elucidated. In the present study, the newly healed skin of the diabetes + FG-4592 group had an irregularly thickened epidermis, with concentrated highly expressed HIF-1α and PCNA+ cells all over the layers. The differentiation of keratinocytes was investigated by WB, and the results showed that the diabetic wound was in a “high differentiation and low proliferation” state, and FG-4592 could reverse this state.

A former study showed that in mouse and human skins, HIF-1α was constitutively expressed in the epidermis, but mainly in the basal layer.[5] Another study used a mouse model of HIF-1α knockout targeted to keratinocytes (K14-Cre/Hif1aflox/flox) and found that loss of epidermal HIF-1α accelerated epidermal aging and affected reepithelialization.[5] This finding supports the view that HIF-1α plays a vital role in keeping keratinocytes young and active.[14] Skin thinning caused by diabetes [Figure 2] is like skin thinning caused by aging to some extent, and both are accompanied by downregulation of HIF-1 signaling. Another study showed that HIF-PHD2 deficiency in keratinocytes promoted wound healing, whereas deficiency in myeloid or endothelial cells did not cause accelerated wound healing,[23] and these findings emphasized the importance of focusing on keratinocytes under the regulation of HIF-1 signaling.

Another recent study reported that Notch1 signaling is activated by hyperglycemia,[24] which is in accordance with the findings of the present study. FG-4592 reversed the hyperactivated Notch1 signaling in diabetic mice. In the authors’ previous study, Notch1 signaling was inhibited after injury in normal wound healing.[25] In most repair processes, cells undergo a fate of “dedifferentiation–proliferation–differentiation.”[26] Notch1 signaling, as a switch regulating proliferation and differentiation in keratinocytes, plays an important role in this process [Figure 6]. Therefore, the inhibition of Notch1 signaling is one of the important mechanisms of roxadustat in promoting the re-epithelialization of diabetic wounds.

This study has some limitations. First, it primarily models type 1 diabetes, so the applicability of the findings to type 2 diabetes is unclear. Studies have shown that hyperglycemia and hyperlipidemia caused by type 2 diabetes mellitus significantly decrease the stability and function of HIF-1a,[2] so targeting HIF-1 signaling pathway therapy may be suitable for type 2 diabetes. Second, while the study provides evidence of roxadustat’s effects on HIF-1α and Notch1 signaling, the underlying mechanisms, particularly how these pathways intersect, could be further elucidated. Further experiments, such as the use of Notch1 signaling pathway inhibitors or siRNA knockdown techniques, could provide deeper insights into the mechanisms by which roxadustat modulates keratinocyte differentiation and angiogenesis. Third, the results of animal experiments cannot be fully applied to clinical practice. Further clinical study is required to identify the optimal dosage, an appropriate dosing interval, and the side effects of FG-4592 to treat diabetic wounds.

SUMMARY

Roxadustat reversed the slow wound healing caused by diabetes and considerably improved the quality of healing. Roxadustat also activated the inhibited HIF-1 signaling in diabetic wounds and inhibited the hyperactivated Notch1 signaling, which kept keratinocytes in a “high proliferation and low differentiation” state. Roxadustat reversed the low expression levels of VEGF and CD31 in diabetic mice and accelerated the wound angiogenesis process. These findings suggest that roxadustat is a potential therapeutic drug that acts as a catalyst for diabetic wound healing through reepithelialization and angiogenesis.

AVAILABILITY OF DATA AND MATERIALS

The data that support the findings of this study are available from the corresponding author upon reasonable request.

ACKNOWLEDGMENT

We thank Professor Yue-sheng Huang for his constructive guidance and help with the ideas and methods of this study.

ABBREVIATIONS

CD31: Platelet endothelial cell adhesion molecule-1

FG-4592: Roxadustat

GAPDH: Glyceraldehyde-3-phosphate dehydrogenase

Glu: Glucose

HE: Hematoxylin & eosin

HIF-1α: hypoxia-inducible factor-1α

HIF-PHD: hypoxia-inducible factor prolyl hydroxylase

IF: Immunofluorescence

IHC: Immunohistochemistry

K1: Keratin 1

K10: Keratin 10

K14: Keratin 14

Mean ± SD: Mean ± Standard deviation

NICD: Notch Intracellular domain

PBS: Phosphate-buffered saline

PCNA: Proliferating cell nuclear antigen

STZ: Streptozotocin

VEGF: Vascular endothelial growth factor

AUTHOR CONTRIBUTIONS

DT: Concepts, design, statistical analysis, manuscript preparation, funding acquisition; QL: Literature search, experimental studies; KX: Concepts, supervision, project administration; QPL: Concepts, writing-reviewing and editing, funding acquisition. All the authors have read and approved the final manuscript. All authors meet the authorship status of ICMJE.

ETHICS APPROVAL AND CONSENT TO PARTICIPATE

This animal experiment followed the requirements of the ethical review system for experimental animal welfare of Wuhan Myhalic Biotechnology Co., Ltd. Animal Ethics Committee. Approval number HLK-20210906-001. Informed consent to participate is not required as this study does not involve human subjects.

CONFLICT OF INTEREST

The authors declare no conflict of interest.

EDITORIAL/PEER REVIEW

To ensure the integrity and highest quality of CytoJournal publications, the review process of this manuscript was conducted under a double-blind model (authors are blinded for reviewers and vice versa) through an automatic online system.

FUNDING: This work was supported by the Hubei Provincial Natural Science Foundation (2021CFB164); Youth Program of National Natural Science Foundation of China (82302818); Postdoctoral Fund of General Hospital of Central Theater Command (BSH017).

References

  1. , , , . Inflammation in obesity, diabetes, and related disorders. Immunity. 2022;55:31-55.
    [CrossRef] [PubMed] [Google Scholar]
  2. , . Hypoxia and hypoxia-inducible factors in diabetes and its complications. Diabetologia. 2021;64:709-16.
    [CrossRef] [PubMed] [Google Scholar]
  3. . The role of keratinocyte function on the defected diabetic wound healing. Int J Burns Trauma. 2021;11:430-41.
    [CrossRef] [Google Scholar]
  4. , , , , . Regulation of wound healing and fibrosis by hypoxia and hypoxiainducible factor-1. Mol Cells. 2014;37:637-43.
    [CrossRef] [PubMed] [Google Scholar]
  5. , , , , , , et al. Constitutive HIF-1α expression in the epidermis fuels proliferation and is essential for effective barrier formation. J Invest Dermatol. 2024;22:S0022-202X(24)02951-8.
    [CrossRef] [PubMed] [Google Scholar]
  6. , . Regulation of glycolysis by the hypoxiainducible factor (HIF): Implications for cellular physiology. J Physiol. 2021;599:23-37.
    [CrossRef] [PubMed] [Google Scholar]
  7. . Hypoxia-inducible factors and diabetes. J Clin Invest. 2020;130:5063-73.
    [CrossRef] [PubMed] [Google Scholar]
  8. , , , , . Dapagliflozin-loaded exosome mimetics facilitate diabetic wound healing by HIF-1α-mediated enhancement of angiogenesis. Adv Healthc Mater. 2023;12:e2202751.
    [CrossRef] [PubMed] [Google Scholar]
  9. , , . Pathogenesis of diabetic complications: Exploring hypoxic niche formation and HIF-1α activation. Biomed Pharmacother. 2024;172:116202.
    [CrossRef] [PubMed] [Google Scholar]
  10. , , , . The possible mechanisms underlying the impairment of HIF-1α pathway signaling in hyperglycemia and the beneficial effects of certain therapies. Int J Med Sci. 2013;10:1412-21.
    [CrossRef] [PubMed] [Google Scholar]
  11. , , , , , , et al. A small molecule HIF-1α stabilizer that accelerates diabetic wound healing. Nat Commun. 2021;12:3363.
    [CrossRef] [PubMed] [Google Scholar]
  12. , , , , , , et al. Roxadustat for anemia in patients with kidney disease not receiving dialysis. N Engl J Med. 2019;381:1001-10.
    [CrossRef] [PubMed] [Google Scholar]
  13. , , , , , , et al. Roxadustat (FG-4592): Correction of anemia in incident dialysis patients. J Am Soc Nephrol. 2016;27:1225-33.
    [CrossRef] [PubMed] [Google Scholar]
  14. , , , , , , et al. FG-4592 accelerates cutaneous wound healing by epidermal stem cell activation via HIF-1α stabilization. Cell Physiol Biochem. 2018;46:2460-70.
    [CrossRef] [PubMed] [Google Scholar]
  15. , , , , . Roxadustat promotes angiogenesis through HIF-1α/VEGF/VEGFR2 signaling and accelerates cutaneous wound healing in diabetic rats. Wound Repair Regen. 2019;27:324-34.
    [CrossRef] [PubMed] [Google Scholar]
  16. , , , , , . Dedifferentiation of human terminally differentiating keratinocytes into their precursor cells induced by basic fibroblast growth factor. Biol Pharm Bull. 2011;34:1037-45.
    [CrossRef] [PubMed] [Google Scholar]
  17. , , , , , , et al. Saccharomyces boulardii exerts renoprotection by modulating oxidative stress, renin angiotensin system and uropathogenic microbiota in a murine model of diabetes. Life Sci. 2022;301:120616.
    [CrossRef] [PubMed] [Google Scholar]
  18. , , . Effect of Salvia miltiorrhiza combined with roxadustat on wound healing of full-thickness skin defects in diabetic rats and its mechanism. Zhonghua Shao Shang Yu Chuang Mian Xiu Fu Za Zhi. 2024;40:380-8.
    [Google Scholar]
  19. , , , , , , et al. Roxadustat promotes hypoxia-inducible factor-1α/vascular endothelial growth factor signalling to enhance random skin flap survival in rats. Int Wound J. 2023;20:3586-98.
    [CrossRef] [PubMed] [Google Scholar]
  20. , . Disturbed hypoxic responses as a pathogenic mechanism of diabetic foot ulcers. Diabetes Metab Res Rev. 2016;32(Suppl 1):179-85.
    [CrossRef] [PubMed] [Google Scholar]
  21. , , , , , , et al. Controlled delivery of HIF-1α via extracellular vesicles with collagen-binding activity for enhanced wound healing. J Control Release. 2025;380:330-47.
    [CrossRef] [PubMed] [Google Scholar]
  22. , , , , , , et al. Comparison of the hydroxylase inhibitor dimethyloxalylglycine and the iron chelator deferoxamine in diabetic and aged wound healing. Plast Reconstr Surg. 2017;139:695e-706.
    [CrossRef] [PubMed] [Google Scholar]
  23. , , , , , , et al. Loss of epithelial hypoxia-inducible factor prolyl hydroxylase 2 accelerates skin wound healing in mice. Mol Cell Biol. 2013;33:3426-38.
    [CrossRef] [PubMed] [Google Scholar]
  24. , , , , , , et al. Triggering of a Dll4-Notch1 loop impairs wound healing in diabetes. P Natl Acad Sci USA. 2019;116:6985-94.
    [CrossRef] [PubMed] [Google Scholar]
  25. , , , , , . Notch1 signaling contributes to hypoxia-induced high expression of integrin β1 in keratinocyte migration. Sci Rep. 2017;7:43926.
    [CrossRef] [PubMed] [Google Scholar]
  26. , , , , , , et al. Dedifferentiation, proliferation, and redifferentiation of adult mammalian cardiomyocytes after ischemic injury. Circulation. 2017;136:834-48.
    [CrossRef] [PubMed] [Google Scholar]
Show Sections